Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.461
Filtrar
1.
Ecotoxicol Environ Saf ; 275: 116278, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38564860

RESUMEN

Due to the rise in temperature and sea level caused by climate change, the detection rate of aflatoxin B1 (AFB1) in food crops has increased dramatically, and the frequency and severity of aflatoxicosis in humans and animals are also increasing. AFB1 has strong hepatotoxicity, causing severe liver damage and even cancer. However, the mechanism of AFB1 hepatotoxicity remains unclear. By integrating network toxicology, molecular docking and in vivo experiments, this research was designed to explore the potential hepatotoxicity mechanisms of AFB1. Thirty-three intersection targets for AFB1-induced liver damage were identified using online databases. PI3K/AKT1, MAPK, FOXO1 signaling pathways, and apoptosis were significantly enriched. In addition, the proteins of ALB, AKT1, PIK3CG, MAPK8, HSP90AA1, PPARA, MAPK1, EGFR, FOXO1, and IGF1 exhibited good affinity with AFB1. In vivo experiments, significant pathological changes occurred in the liver of mice. AFB1 induction increased the expression levels of EGFR, ERK, and FOXO1, and decreased the expression levsls of PI3K and AKT1. Moreover, AFB1 treatment caused an increase in Caspase3 expression, and a decrease in Bcl2/Bax ratio. By combining network toxicology with in vivo experiments, this study confirms for the first time that AFB1 promotes the FOXO1 signaling pathway by inactivating PI3K/AKT1 and activating EGFR/ERK signaling pathways, hence aggravating hepatocyte apoptosis. This research provides new strategies for studying the toxicity of environmental pollutants and new possible targets for the development of hepatoprotective drugs.


Asunto(s)
Aflatoxina B1 , Enfermedad Hepática Inducida por Sustancias y Drogas , Humanos , Ratones , Animales , Simulación del Acoplamiento Molecular , Aflatoxina B1/toxicidad , Hígado/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores ErbB/metabolismo
2.
Onderstepoort J Vet Res ; 91(1): e1-e6, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38572889

RESUMEN

Global aflatoxin contamination of agricultural commodities is of the most concern in food safety and quality. This study investigated the hepatoprotective effect of 80% methanolic leaf extract of Annona senegalensis against aflatoxin B1 (AFB1)-induced toxicity in rats. A. senegalensis has shown to inhibit genotoxicity of aflatoxin B1 in vitro. The rats were divided into six groups including untreated control, aflatoxin B1 only (negative control); curcumin (positive control; 10 mg/kg); and three groups receiving different doses (100 mg/kg, 200 mg/kg, and 300 mg/kg) of A. senegalensis extract. The rats received treatment (with the exception of untreated group) for 7 days prior to intoxication with aflatoxin B1. Serum levels of aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, lactate dehydrogenase, and creatinine were measured. Hepatic tissues were analysed for histological alterations. Administration of A. senegalensis extract demonstrated hepatoprotective effects against aflatoxin B1-induced toxicity in vivo by significantly reducing the level of serum aspartate aminotransferase and alanine aminotransferase and regenerating the hepatocytes. No significant changes were observed in the levels of alkaline phosphatase, lactate dehydrogenase, and creatinine for the AFB1 intoxicated group, curcumin+AFB1 and Annona senegalensis leaf extract (ASLE)+AFB1 (100 mg/kg, 200 mg/kg, and 300 mg/kg body weight [b.w.]) treated groups. Annona senegalensis is a good candidate for hepatoprotective agents and thus its use in traditional medicine may at least in part be justified.Contribution: The plant extract investigated in this study can be used in animal health to protect the organism from toxicity caused by mycotoxins.


Asunto(s)
Annona , Curcumina , Ratas , Animales , Aflatoxina B1/toxicidad , Curcumina/farmacología , Alanina Transaminasa/farmacología , Fosfatasa Alcalina/farmacología , Creatinina/farmacología , Hígado , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Aspartato Aminotransferasas/farmacología , Lactato Deshidrogenasas
3.
Proc Natl Acad Sci U S A ; 121(16): e2314426121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38574017

RESUMEN

Epstein-Barr Virus (EBV) infects more than 90% of the adult population worldwide. EBV infection is associated with Burkitt lymphoma (BL) though alone is not sufficient to induce carcinogenesis implying the involvement of co-factors. BL is endemic in African regions faced with mycotoxins exposure. Exposure to mycotoxins and oncogenic viruses has been shown to increase cancer risks partly through the deregulation of the immune response. A recent transcriptome profiling of B cells exposed to aflatoxin B1 (AFB1) revealed an upregulation of the Chemokine ligand 22 (CCL22) expression although the underlying mechanisms were not investigated. Here, we tested whether mycotoxins and EBV exposure may together contribute to endemic BL (eBL) carcinogenesis via immunomodulatory mechanisms involving CCL22. Our results revealed that B cells exposure to AFB1 and EBV synergistically stimulated CCL22 secretion via the activation of Nuclear Factor-kappa B pathway. By expressing EBV latent genes in B cells, we revealed that elevated levels of CCL22 result not only from the expression of the latent membrane protein LMP1 as previously reported but also from the expression of other viral latent genes. Importantly, CCL22 overexpression resulting from AFB1-exposure in vitro increased EBV infection through the activation of phosphoinositide-3-kinase pathway. Moreover, inhibiting CCL22 in vitro and in humanized mice in vivo limited EBV infection and decreased viral genes expression, supporting the notion that CCL22 overexpression plays an important role in B cell infection. These findings unravel new mechanisms that may underpin eBL development and identify novel pathways that can be targeted in drug development.


Asunto(s)
Linfoma de Burkitt , Infecciones por Virus de Epstein-Barr , Animales , Ratones , Herpesvirus Humano 4/genética , Infecciones por Virus de Epstein-Barr/complicaciones , Aflatoxina B1/toxicidad , Ligandos , Linfoma de Burkitt/metabolismo , Quimiocinas , Carcinogénesis
4.
Food Funct ; 15(7): 3615-3628, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38470843

RESUMEN

Aflatoxin B1 (AFB1) causes serious immunotoxicity and has attracted considerable attention owing to its high sensitivity and common chemical-viral interactions in living organisms. However, the sensitivity of different species to AFB1 widely varies, which cannot be explained by the different metabolism in species. The gut microbiota plays a crucial role in the immune system, but the interaction of the microbiota with AFB1-induced immunotoxicity still needs to be determined. Our results indicated that AFB1 exposure disrupted the structure of the gut microbiota and damaged the gut barrier, which caused translocation of microbiota metabolites, lipopolysaccharides, to the spleen. Subsequently, pyroptosis of the spleen was activated. Interestingly, AFB1 exposure had little effect on the splenic pyroptosis of pseudo-germfree mice (antibiotic mixtures eliminated their gut microbiota, ABX). Then, fecal microbiota transplant (FMT) and sterile fecal filtrate (SFF) were employed to validate the function of the gut microbiota and its metabolites in AFB1-induced splenic pyroptosis. The AFB1-disrupted microbiota and its metabolites significantly promoted splenic pyroptosis, which was worse than that in control mice. Overall, AFB1-induced splenic pyroptosis is associated with the gut microbiota and its metabolites, which was further demonstrated by FMT and SFF. The mechanism of AFB1-induced splenic pyroptosis was explored for the first time, which paves a new way for preventing and treating the immunotoxicity from mycotoxins by regulating the gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Piroptosis , Animales , Ratones , Aflatoxina B1/toxicidad , Bazo , Heces
5.
Food Chem ; 447: 138909, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38489879

RESUMEN

The purpose of this review was to investigate the current knowledge about aflatoxin B1 (AFB1) and ochratoxin A (OTA) toxicity and the possible beneficial role of bioactive compounds by using in vitro and in vivo models. Although AFB1 and OTA were tested in a similar percentage, the majority of studies focused on nephrotoxicity, hepatotoxicity, immune toxicity and neurotoxicity in which oxidative stress, inflammation, structural damage and apoptosis were the main mechanisms of action reported. Conversely, several biological compounds were assayed in order to modulate mycotoxins damage mainly in the liver, brain, kidney and immune system. Among them, pumpkin, curcumin and fermented whey were the most employed. Although a clear progress has been made by using in vivo models, further research is needed to assess not only the toxicity of multiple mycotoxins contamination but also the effect of functional compounds mixture, thereby reproducing more realistic situations for human health risk assessment.


Asunto(s)
Micotoxinas , Ocratoxinas , Humanos , Aflatoxina B1/toxicidad , Ocratoxinas/toxicidad , Micotoxinas/toxicidad , Hígado
6.
J Hazard Mater ; 469: 133916, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38479137

RESUMEN

Aflatoxins from the fungus Aspergillus flavus (A. flavus) that contaminate stored peanuts is a major hazard to human health worldwide. Reducing A. flavus in soil can decrease the risk of aflatoxins in stored peanuts. In this experiment, we determined whether peanuts grown on soil fumigated with dazomet (DZ), metham sodium (MS), allyl isothiocyanate (AITC), chloropicrin (PIC) or dimethyl disulfide (DMDS) would reduce of the quantity of A. flavus and its toxin's presence. The results of bioassays and field tests showed that PIC was the most effective fumigant for preventing and controlling A. flavus, followed by MS. PIC and MS applied to the soil for 14 d resulted in LD50 values against A. flavus of 3.558 and 4.893 mg kg-1, respectively, leading to almost 100% and 98.82% effectiveness of A. flavus, respectively. Peanuts harvested from fumigated soil and then stored for 60 d resulted in undetectable levels of aflatoxin B1 (AFB1) compared to unfumigated soil that contained 0.64 ug kg-1 of AFB1, which suggested that soil fumigation can reduce the probability of aflatoxin contamination during peanut storage and showed the potential to increase the safety of peanuts consumed by humans. Further research is planned to determine the practical value of our research in commercial practice.


Asunto(s)
Aflatoxina B1 , Aflatoxinas , Humanos , Aflatoxina B1/toxicidad , Aflatoxina B1/análisis , Arachis , Suelo , Desinfección , Aspergillus flavus , Aflatoxinas/toxicidad , Aflatoxinas/análisis
7.
Arch Toxicol ; 98(4): 1081-1093, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38436695

RESUMEN

Large interspecies differences between rats and mice concerning the hepatotoxicity and carcinogenicity of aflatoxin B1 (AFB1) are known, with mice being more resistant. However, a comprehensive interspecies comparison including subcellular liver tissue compartments has not yet been performed. In this study, we performed spatio-temporal intravital analysis of AFB1 kinetics in the livers of anesthetized mice and rats. This was supported by time-dependent analysis of the parent compound as well as metabolites and adducts in blood, urine, and bile of both species by HPLC-MS/MS. The integrated data from intravital imaging and HPLC-MS/MS analysis revealed major interspecies differences between rats and mice: (1) AFB1-associated fluorescence persisted much longer in the nuclei of rat than mouse hepatocytes; (2) in the sinusoidal blood, AFB1-associated fluorescence was rapidly cleared in mice, while a time-dependent increase was observed in rats in the first three hours after injection followed by a plateau that lasted until the end of the observation period of six hours; (3) this coincided with a far stronger increase of AFB1-lysine adducts in the blood of rats compared to mice; (4) the AFB1-guanine adduct was detected at much higher concentrations in bile and urine of rats than mice. In both species, the AFB1-glutathione conjugate was efficiently excreted via bile, where it reached concentrations at least three orders of magnitude higher compared to blood. In conclusion, major differences between mice and rats were observed, concerning the nuclear persistence, formation of AFB1-lysine adducts, and the AFB1-guanine adducts.


Asunto(s)
Aflatoxinas , Ratas , Ratones , Animales , Aflatoxinas/metabolismo , Aflatoxinas/toxicidad , Lisina/metabolismo , 60705 , Espectrometría de Masas en Tándem , Hígado/metabolismo , Aflatoxina B1/toxicidad , Guanina/metabolismo , Microscopía Intravital
8.
Sci Total Environ ; 927: 171973, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38547995

RESUMEN

The aim of this study was to investigate the alleviating effect of selenomethionine (SeMet) on aflatoxin B1 (AFB1)-induced testicular injury in rabbits. Twenty-five 90-d-old rabbits were randomly divided into 5 groups (the control group, the AFB1 group, the 0.2 mg/kg SeMet + AFB1 group, the 0.4 mg/kg SeMet + AFB1 group and the 0.6 mg/kg SeMet + AFB1 group). After 1 d of the experiment, the SeMet-treated groups were fed 0.2 mg/kg SeMet, 0.4 mg/kg SeMet, or 0.6 mg/kg SeMet daily, and the remaining two groups were fed a normal diet for 30 d. On Day 31, all rabbits in the model group and the three treatment groups were fed 0.5 mg/kg AFB1 for 21 d. The levels of testosterone (T), luteinizing hormone (LH) and follicle stimulating hormone (FSH) in rabbit plasma were detected. Rabbit semen was collected, and its quality was evaluated. Pathological changes in rabbit testes were observed by hematoxylin-eosin (HE) staining. The expression of related proteins in testicular tissue was detected by immunohistochemistry, immunofluorescence and western blot (WB) analysis. Enzyme-linked immunosorbent assays (ELISAs) were used to detect oxidative stress-related indices and inflammatory factors in testicular tissue. The results showed that AFB1 can induce oxidative stress and inflammation to activate the p38/MSK/NF-κB signalling pathway, mediate apoptosis, inhibit the proliferation and differentiation of testicular cells, destroy the integrity of the blood-testis barrier (BTB) and the normal structure of the testis, and reduce the content of sex hormones and semen quality. SeMet pretreatment significantly alleviated testicular injury oxidative stress, and the inflammatory response in rabbits. Thus, we demonstrated that SeMet restores AFB1-induced testicular toxicity by inhibiting the p38/MSK/NF-κB signalling pathway. In addition, in this study, 0.4 mg/kg SeMet had the most impactful effect.


Asunto(s)
Aflatoxina B1 , Selenometionina , Testículo , Animales , Masculino , Conejos , Aflatoxina B1/toxicidad , Selenometionina/farmacología , Testículo/efectos de los fármacos , Testosterona/sangre , Sustancias Protectoras/farmacología , Enfermedades Testiculares/prevención & control , Enfermedades Testiculares/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Hormona Luteinizante/sangre , Apoptosis/efectos de los fármacos
9.
Mutagenesis ; 39(3): 181-195, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38468450

RESUMEN

Aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are mycotoxins widely distributed in maize and maized-based products, often occurring together. The implications of co-exposure to aflatoxin and fumonsin for human health are numerous, but a particular concern is the potential of FB1 to modulate AFB1 hepatotoxicity. This study evaluated the toxicity of these mycotoxins, alone or combined, in a human non-tumorigenic liver cell line, HHL-16 cells, and assessed the effects of AFB1 and FB1 on expression of genes involved in immune and growth factor pathways. The results demonstrated that in HHL-16 cells, both AFB1 and FB1 had dose-dependent and time-dependent toxicity, and the combination of them showed a synergistic toxicity in the cells. Moreover, AFB1 caused upregulation of IL6, CCL20, and BMP2, and downregulation of NDP. In combination of AFB1 with FB1, gene expression levels of IL6 and BMP2 were significantly higher compared to individual FB1 treatment, and had a tendency to be higher than individual AFB1 treatment. This study shows that FB1 may increase the hepatoxicity of AFB1 through increasing the inflammatory response and disrupting cell growth pathways.


Asunto(s)
Aflatoxina B1 , Fumonisinas , Hepatocitos , Fumonisinas/toxicidad , Humanos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Aflatoxina B1/toxicidad , Línea Celular , Inflamación/genética , Inflamación/inducido químicamente , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo
10.
BMC Vet Res ; 20(1): 108, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38500117

RESUMEN

BACKGROUND: Camel milk and silymarin have many different beneficial effects on several animal species. Meanwhile, Aflatoxins are mycotoxins with extraordinary potency that pose major health risks to several animal species. Additionally, it has been documented that aflatoxins harm the reproductive systems of a variety of domestic animals. The present design aimed to investigate the impact of aflatoxin B1 (AFB1) on rat body weight and reproductive organs and the ameliorative effects of camel milk and silymarin through measured serum testosterone, testes pathology, and gene expression of tumor necrosis factor (TNF-α), luteinizing hormone receptor (LHR), and steroidogenic acute regulatory protein (StAR) in the testes. A total of sixty mature male Wister white rats, each weighing an average of 83.67 ± 0.21 g, were used. There were six groups created from the rats. Each division had ten rats. The groups were the control (without any treatment), CM (1 ml of camel milk/kg body weight orally), S (20 mg silymarin/kg b. wt. suspension, orally), A (1.4 mg aflatoxin/kg diet), ACM (aflatoxin plus camel milk), and AS (aflatoxin plus silymarin). RESULTS: The results indicated the positive effects of camel milk and silymarin on growth, reproductive organs, and gene expression of TNF-α, LHR, and StAR with normal testicular architecture. Also, the negative effect of AFB1 on the rat's body weight and reproductive organs, as indicated by low body weight and testosterone concentration, was confirmed by the results of histopathology and gene expression. However, these negative effects were ameliorated by the ingestion of camel milk and silymarin. CONCLUSION: In conclusion, camel milk and silymarin could mitigate the negative effect of AFB1 on rat body weight and reproductive organs.


Asunto(s)
Aflatoxinas , Silimarina , Masculino , Ratas , Animales , Aflatoxina B1/toxicidad , Aflatoxina B1/metabolismo , Silimarina/farmacología , Camelus , Leche , Factor de Necrosis Tumoral alfa/metabolismo , Ratas Wistar , Testículo/metabolismo , Testosterona/metabolismo , Peso Corporal
11.
Int J Mol Sci ; 25(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38474096

RESUMEN

Aflatoxins are harmful natural contaminants found in foods and are known to be hepatotoxic. However, recent studies have linked chronic consumption of aflatoxins to nephrotoxicity in both animals and humans. Here, we conducted a systematic review of active compounds, crude extracts, herbal formulations, and probiotics against aflatoxin-induced renal dysfunction, highlighting their mechanisms of action in both in vitro and in vivo studies. The natural products and dietary supplements discussed in this study alleviated aflatoxin-induced renal oxidative stress, inflammation, tissue damage, and markers of renal function, mostly in animal models. Therefore, the information provided in this review may improve the management of kidney disease associated with aflatoxin exposure and potentially aid in animal feed supplementation. However, future research is warranted to translate the outcomes of this study into clinical use in kidney patients.


Asunto(s)
Aflatoxinas , Productos Biológicos , Enfermedades Renales , Animales , Humanos , Aflatoxinas/toxicidad , Aflatoxina B1/toxicidad , Suplementos Dietéticos
12.
J Agric Food Chem ; 72(11): 5966-5974, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38446589

RESUMEN

Mycotoxin contamination is an important issue for food safety and the environment. Removing mycotoxins from food without losing nutrients and flavor components remains a challenge. In this study, a novel strategy was proposed for the targeted removal of aflatoxin B1 (AFB1) from peanut oil using an amphipathic enzyme-metal hybrid nanoreactor (PL-GOx-Fe3O4@COF) constructed with covalent organic frameworks (COFs) which can selectively adsorb AFB1. Due to the confined space provided by COFs and the proximity effect between GOx and Fe3O4, the detoxification of AFB1 is limited in the nanoreactor without affecting the composition and properties of the oil. The detoxification efficiency of AFB1 in the chemoenzymatic cascade reaction catalyzed by PL-GOx-Fe3O4@COF is six times higher than that of the combination of free GOx and Fe3O4. The AFB1 transformation product has nontoxicity to kidney and liver cells. This study provides a powerful tool for the targeted removal of mycotoxins from edible oils.


Asunto(s)
Aflatoxina B1 , Inocuidad de los Alimentos , Aflatoxina B1/toxicidad , Hepatocitos , Aceite de Cacahuete , Nanotecnología
13.
J Agric Food Chem ; 72(11): 5955-5965, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38451160

RESUMEN

The worldwide prevalence of Aflatoxin B1 (AFB1), which contaminates feedstock and food, is on the rise. AFB1 inhibits testosterone (T) biosynthesis, but the mechanism is not yet clear. By establishing in vivo and in vitro models, this study found the number of Leydig cells (LCs), T content, and the expression of T biosynthesis key enzymes were suppressed after AFB1 treatment. AFB1 exposure also increased reactive oxygen species (ROS) and promoted mitochondrial injury and mitochondrial pathway apoptosis. Moreover, the AMPK signaling pathway was activated, and using an AMPK inhibitor relieved apoptosis and the suppressed T biosynthesis key enzymes of LCs caused by AFB1 through regulating downstream p53 and Nur77. Additionally, adding ROS intervention could inhibit AMPK activation and alleviate the decreased T content caused by AFB1. In summary, AFB1 promotes the apoptosis of LCs and inhibits T biosynthesis key enzyme expression via activating the ROS/AMPK signaling pathway, which eventually leads to T synthesis disorder.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Aflatoxina B1 , Ratones , Masculino , Animales , Especies Reactivas de Oxígeno/metabolismo , Aflatoxina B1/toxicidad , Aflatoxina B1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Transducción de Señal , Testosterona , Apoptosis , Estrés Oxidativo
14.
Chem Biol Interact ; 392: 110923, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38382706

RESUMEN

Aflatoxin B1 (AFB1) is the most toxic mycotoxin and a proven human carcinogen that requires metabolic activation, known by cytochrome P450 (CYP) 1A2 and 3A4. Previous evidence showed that AFB1 is activated by human recombinant CYP1A1 expressed in budding yeast. Yet, the toxicity, in particular the genotoxicity of the reactive metabolites formed from AFB1 remains unclear. Humans could be exposed to both AFB1 and benzo(a)pyrene (BaP) simultaneously, thus we were interested in their combined genotoxic effects subsequent to metabolic activation by CYP1A1. In this study, molecular docking of AFB1 to human CYP1A1 indicated that AFB1 is valid as a substrate. In the incubations with AFB1 in human CYP1A1-expressed microsomes, AFM1 as a marking metabolite of AFB1 was detected. Moreover, AFB1 induced micronucleus formation in a Chinese hamster V79-derived cell line and in a human lung epithelial BEAS-2B cell line, both expressing recombinant human CYP1A1, V79-hCYP1A1 and 2B-hCYP1A1 cells, respectively. Immunofluorescence of centromere protein B stained micronuclei was dominant in AFB1-treated BEAS-2B cells exposed to AFB1, suggesting an aneugenic effect. Moreover, AFB1 elevated the levels of ROS, 8-OHdG, AFB1-DNA adduct, and DNA breaks in 2B-hCYP1A1 cells, compared with those in the parental BEAS-2B cells. Meanwhile, AFB1 increased CYP1A1, RAD51, and γ-H2AX protein levels in 2B-hCYP1A1 cells, which were attenuated by the CYP1A1 inhibitor bergamottin. Co-exposure of AFB1 with BaP increased 8-OHdG, RAD51, and γ-H2AX levels (indicating DNA damage). In conclusion, AFB1 could be activated by human CYP1A1 for potent aneugenicity, which may be further enhanced by co-exposure to BaP.


Asunto(s)
Citocromo P-450 CYP1A1 , Sistema Enzimático del Citocromo P-450 , Animales , Humanos , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Aflatoxina B1/toxicidad , Aflatoxina B1/metabolismo , Benzo(a)pireno/toxicidad , Aneugénicos , Simulación del Acoplamiento Molecular , Mamíferos/metabolismo
15.
J Hazard Mater ; 468: 133836, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38394902

RESUMEN

Global aflatoxin B1 (AFB1) contamination is inevitable, and it can significantly damage testicular development. However, the current mechanism is confusing. Here, by integrating the transcriptome, microbiome, and serum metabolome, we comprehensively explain the impact of AFB1 on testis from the gut-metabolism-testis axis. Transcriptome analysis suggested that AFB1 exposure directly causes abnormalities in testicular inflammation-related signalling, such as tumor necrosis factor (TNF) pathway, and proliferation-related signalling pathways, such as phosphatidylinositide 3-kinases-protein kinase B (PI3K-AKT) pathway, which was verified by immunofluorescence. On the other hand, we found that upregulated inflammatory factors in the intestine after AFB1 exposure were associated with intestinal microbial dysbiosis, especially the enrichment of Bacilli, and enrichment analysis showed that this may be related to NLR family pyrin domain containing 3 (NLRP3)-mediated NOD-like receptor signalling. Also, AFB1 exposure caused blood metabolic disturbances, manifested as decreased hormone levels and increased oxidative stress. Significantly, B. licheniformis has remarkable AFB1 degradation efficiency (> 90%). B. licheniformis treatment is effective in attenuating gut-testis axis damage caused by AFB1 exposure through the above-mentioned signalling pathways. In conclusion, our findings indicate that AFB1 exposure disrupts testicular development through the gut-metabolism-testis axis, and B. licheniformis can effectively degrade AFB1.


Asunto(s)
Bacillus licheniformis , Testículo , Masculino , Humanos , Aflatoxina B1/toxicidad , Aflatoxina B1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Metaboloma
16.
Mycotoxin Res ; 40(2): 269-277, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38421516

RESUMEN

This experiment was conducted to determine the effect of an adsorbent material based on powdered alfalfa leaves added in the aflatoxin B1 (AFB1)-contaminated diet of turkey poults on production parameters, blood cell count, serum biochemistry, liver enzymes, and liver histology. For this purpose, three hundred and fifty female Nicholas-700 poults were randomly assigned into five treatments: (1) Control, AFB1-free diet; (2) AF, diet contaminated with 250 ng AFB1/g; (3) Alfalfa, AFB1-free diet + 0.5% (w/w) adsorbent; (4) AF+alfalfa, diet contaminated with 250 ng AFB1/g + 0.5% (w/w) adsorbent, and (5) AF+ yeast cell wall (YCW), diet contaminated with 250 ng AFB1/g + 0.5% (w/w) of yeast cell wall (a commercial mycotoxin binder used as reference material). The in vivo efficacy of powdered alfalfa leaves was assessed during a 28-day period. In general, the addition of powdered alfalfa leaves in the AFB1-free diet gave the best performance results (body weight, body weight gain, and feed intake) and improved the values of total protein, glucose, calcium, creatinine, and blood urea nitrogen. Moreover, the addition of powdered alfalfa leaves in the AFB1-contaminated diet enhanced body weight and body weight gain and significantly reduced the feed intake, compared to the AF and AF+YCW groups. Additionally, significant alterations in serum parameters were observed in poults intoxicated with the AFB1, compared to the Control group. Furthermore, typical histopathological lesions were observed in the liver of the AF group, which were significantly ameliorated with the addition of powdered alfalfa leaves. Conclusively, these results pointed out that low inclusion of powdered alfalfa leaves in the contaminated feed counteracted the adverse effects of AFB1 in turkey poults.


Asunto(s)
Aflatoxina B1 , Alimentación Animal , Medicago sativa , Hojas de la Planta , Pavos , Animales , Aflatoxina B1/toxicidad , Medicago sativa/química , Hojas de la Planta/química , Alimentación Animal/análisis , Femenino , Hígado/efectos de los fármacos , Hígado/patología , Dieta/veterinaria , Polvos , Peso Corporal/efectos de los fármacos
17.
Mycotoxin Res ; 40(2): 255-268, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38400893

RESUMEN

Aflatoxin B1 (AFB1) is a widespread toxic contamination in feed for animals. The primary active component of turmeric, curcumin (Cur), is an antioxidant and an anti-inflammatory. However, it is yet unknown how AFB1 affects the intestinal epithelial barrier and whether Cur acts as a protective mechanism when exposed to AFB1. Here, we explored the mechanism of AFB1-induced intestinal injury from intestinal epithelial barrier, inflammation, pyroptosis, and intestinal flora, and evaluated the protective role of Cur. We found that AFB1 caused weight loss and intestinal morphological damage that is mainly characterized by shortened intestinal villi, deepened crypts, and damaged intestinal epithelium. Exposure to AFB1 decreased the expression of Claudin-1, MUC2, ZO-1, and Occludin and increased the expression of pyroptosis-related factors (NLRP3, GSDMD, Caspase-1, IL-1ß, and IL-18) and inflammation-related factors (TLR4, NF-κB, IκB, IFN-γ, and TNF-α). Furthermore, ileal gut microbiota was altered, and simultaneously, the Lactobacillus abundance was decreased. The gut microbiota interacts with a wide range of physiologic functions and disease development in the host through its metabolites, and disturbances in gut microbial metabolism can cause functional impairment of the ileum. Meanwhile, Cur can ameliorate histological ileum injuries and intestinal flora disturbance caused by AFB1. We found that Cur reversed the effects of AFB1 through modulating both NLRP3 inflammasome and the TLR4/NF-κB signaling pathway. In conclusion, AFB1 can induce inflammatory damage and pyroptosis in duck ileum, while Cur has obviously protective effects on all the above damages.


Asunto(s)
Aflatoxina B1 , Curcumina , Patos , Íleon , Inflamasomas , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Receptor Toll-Like 4 , Animales , Aflatoxina B1/toxicidad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Curcumina/farmacología , Inflamasomas/metabolismo , Íleon/efectos de los fármacos , Íleon/patología , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología
18.
Environ Pollut ; 345: 123474, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38309422

RESUMEN

Aflatoxins (AFTs), a type of mycotoxin mainly produced by Aspergillus parasiticus and Aspergillus flavus, could be detected in food, feed, Chinese herbal medicine, grain crops and poses a great threat to public health security. Among them, aflatoxin B1 (AFB1) is the most toxic one. Exposure to AFB1 poses various health risks to both humans and animals, including the development of chronic inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, and cancer. The molecular mechanisms underlying these risks are intricate and dependent on specific contexts. This review primarily focuses on summarizing the protective effects of quercetin, a natural phenolic compound, in mitigating the toxic effects induced by AFB1 in both in vitro experiments and animal models. Additionally, the review explores the molecular mechanisms that underlie these protective effects. Quercetin has been demonstrated to not only have the direct inhibitory action on the production of AFTs from Aspergillus, both also possess potent ameliorative effects against AFB1-induced cytotoxicity, hepatotoxicity, and neurotoxicity. These effects are attributed to the inhibition of oxidative stress, mitochondrial dysfunction, mitochondrial apoptotic pathway, and inflammatory response. It could also directly target several metabolic enzymes (i.e., CYP3As and GSTA1) to reduce the production of toxic metabolites of AFB1 within cells, then reduce AFB1-induced cytotoxicity. In conclusion, this review highlights quercetin is a promising detoxification agent for AFB1. By advancing our understanding of the protective mechanisms offered by quercetin, we aim to contribute to the development of effective detoxification agents against AFB1, ultimately promoting better health outcomes.


Asunto(s)
Aflatoxina B1 , Quercetina , Animales , Humanos , Aflatoxina B1/toxicidad , Quercetina/farmacología , Estrés Oxidativo , Fenoles/farmacología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico
19.
Toxicon ; 240: 107640, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38325757

RESUMEN

The effects of dietary supplementation with Capsicum annuum fruit pericarp powder (CPP) and Capsicum annuum fruit seed powder (CSP) on the health and performance of broiler chickens exposed to aflatoxin B1 (AFB1) was investigated. Four dietary groups were established: CON (control), AFT (0.5 mg/kg AFB1), CPAF (0.5 g/kg CPP and 0.5 mg/kg AFB1), and CSAF (0.5 g/kg CSP and 0.5 mg/kg AFB1). The AFT group shows a significant (P < 0.05) reduction in the relative growth rate compared to CON, CPAF, and CSAF. In contrast, the latter two groups exhibit growth rates similar (P > 0.05) to CON. Additionally, immunoglobulin levels (IgG, IgM, and IgA) in the AFT group are significantly (P < 0.05) lower compared to the other treatment groups. Serum interleukin-6 levels in the CPAF and CSAF groups were similar (P > 0.05) to CON but higher (P < 0.05) than in AFT. Tumor necrosis factor-alpha levels were elevated (P < 0.05) in AFT compared to the other treatment groups. Interferon-gamma concentrations in AFT were significantly (P < 0.05) lower than in the other treatment groups. The liver histology reveals that the AFT treatment group has periportal hepatic inflammation. In contrast, the CPAF and CSAF treatment groups exhibit normal hepatic microanatomy. In conclusion, 0.5 g/kg CPAF dietary supplementation may help to ameliorate the adverse effects of AFB1 exposure on broiler chicken health, specifically the growth, immune parameters and liver histology.


Asunto(s)
Capsicum , Factor de Activación Plaquetaria/análogos & derivados , Animales , Pollos , Aflatoxina B1/toxicidad , Aflatoxina B1/análisis , Polvos/farmacología , Citocinas , Adipoquinas/farmacología , Hígado , Suplementos Dietéticos , Inmunoglobulinas , Carne , Alimentación Animal/análisis
20.
Ecotoxicol Environ Saf ; 272: 116049, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38301584

RESUMEN

Global concern exists regarding the contamination of food and animal feed with aflatoxin B1 (AFB1), which poses a threat to the health of both humans and animals. Previously, we found that a laccase from Bacillus subtilis (BsCotA) effectively detoxified AFB1 in a reaction mediated by methyl syringate (MS), although the underlying mechanism has not been determined. Therefore, our primary objective of this study was to explore the detoxification mechanism employed by BsCotA. First, the enzyme and mediator dependence of AFB1 transformation were studied using the BsCotA-MS system, which revealed the importance of MS radical formation during the oxidation process. Aflatoxin Q1 (AFQ1) resulting from the direct oxidation of AFB1 by BsCotA, was identified using ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). The results of UPLC-MS/MS and density functional theory calculations indicated that the products included AFQ1, AFB1-, and AFD1-MS-coupled products in the BsCotA-MS system. The toxicity evaluations revealed that the substances derived from the transformation of AFB1 through the BsCotA-MS mechanism exhibited markedly reduced toxicity compared to AFB1. Finally, we proposed a set of different AFB1-transformation pathways generated by the BsCotA-MS system based on the identified products. These findings greatly enhance the understanding of the AFB1-transformation mechanism of the laccase-mediator system.


Asunto(s)
Aflatoxina B1 , Ácido Gálico/análogos & derivados , Lacasa , Humanos , Aflatoxina B1/toxicidad , Aflatoxina B1/química , Cromatografía Liquida , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...